Pectoralis major muscle atrophy is associated with mitochondrial energy wasting in cachectic patients with gastrointestinal cancer

Adeline Dolly, Thierry Lecomte, Nicolas Tabchouri, Morgane Caulet, Nicolas Michot, Benjamin Anon, Romain Chautard, Yoann Desvignes, Mehdi Ouaissi, Gaëlle Fromont-Hankard, Jean-François Dumas, Stéphane Servais, Adeline Dolly, Thierry Lecomte, Nicolas Tabchouri, Morgane Caulet, Nicolas Michot, Benjamin Anon, Romain Chautard, Yoann Desvignes, Mehdi Ouaissi, Gaëlle Fromont-Hankard, Jean-François Dumas, Stéphane Servais

Abstract

Background: Cancer cachexia is a multifactorial syndrome characterized by involuntary and pathological weight loss, mainly due to skeletal muscle wasting, resulting in a decrease in patients' quality of life, response to cancer treatments, and survival. Our objective was to investigate skeletal muscle alterations in cachectic cancer patients.

Methods: This is a prospective study of patients managed for pancreatic or colorectal cancer with an indication for systemic chemotherapy (METERMUCADIG - NCT02573974). One lumbar CT image was used to determine body composition. Patients were divided into three groups [8 noncachectic (NC), 18 with mild cachexia (MC), and 19 with severe cachexia (SC)] based on the severity of weight loss and muscle mass. For each patient, a pectoralis major muscle biopsy was collected at the time of implantable chamber placement. We used high-resolution oxygraphy to measure mitochondrial muscle oxygen consumption on permeabilized muscle fibres. We also performed optical and electron microscopy analyses, as well as gene and protein expression analyses.

Results: Forty-five patients were included. Patients were 67% male, aged 67 years (interquartile range, 59-77). Twenty-three (51%) and 22 (49%) patients were managed for pancreatic and colorectal cancer, respectively. Our results show a positive correlation between median myofibres area and skeletal muscle index (P = 0.0007). Cancer cachexia was associated with a decrease in MAFbx protein expression (P < 0.01), a marker of proteolysis through the ubiquitin-proteasome pathway. Mitochondrial oxygen consumption related to energy wasting was significantly increased (SC vs. NC, P = 0.028) and mitochondrial area tended to increase (SC vs. MC, P = 0.056) in SC patients. On the contrary, mitochondria content and networks remain unaltered in cachectic cancer patients. Finally, our results show no dysfunction in lipid storage and endoplasmic reticulum homeostasis.

Conclusions: This clinical protocol brings unique data that provide new insight to mechanisms underlying muscle wasting in cancer cachexia. We report for the first time an increase in mitochondrial energy wasting in the skeletal muscle of severe cachectic cancer patients. Additional clinical studies are essential to further the exploring and understanding of these alterations.

Keywords: Cancer cachexia; Clinical study; Mitochondrial bioenergetics; Myosteatosis; Pectoralis major; Proteolysis.

Conflict of interest statement

The authors declare that they have no conflict of interest in connection with this article.

© 2022 The Authors. Journal of Cachexia, Sarcopenia and Muscle published by John Wiley & Sons Ltd on behalf of Society on Sarcopenia, Cachexia and Wasting Disorders.

Figures

Figure 1
Figure 1
Pectoralis major myofibres area decreases with progression of cachexia in gastrointestinal cancer patients. (A) Representative observation by optical microscopy of pectoralis major myofibres stained with toluidine blue in noncachectic patients (NC), patients with mild cachexia (MC) or severe cachexia (SC). Scale 100 μm. (B) Median myofibres area. For each patient, the median myofibres area was calculated on approximately 200 myofibres. Correlations between median myofibres area and (C) skeletal muscle index (SMI) (Spearman); (D) body weight loss percentage (Pearson). (E) Median area of myofibrils observed by transmission electron microscopy. For each patient, four cross‐sections, each on a different myofibre, were used. Median myofibrils area was calculated on at least 30 myofibrils per section. Data are presented with IQR box and min‐to‐max whiskers and were compared using Kruskal–Wallis tests with Dunn's post hoc tests. Outliers were excluded using Grubbs' test α = 0.05. Men: black dots, women: white triangles. *P < 0.05, **P < 0.01.
Figure 2
Figure 2
Cancer cachexia is associated with decrease in MAFbx protein expression in pectoralis major muscle. (A) Protein expression levels of two markers of the proteasome‐mediated degradation pathway: MuRF1 and MAFbx, measured in pectoralis major biopsies from noncachectic patients (NC), patients with mild cachexia (MC), or severe cachexia (SC). (B) Spearman correlation between the protein expression level of MAFbx and the skeletal muscle index (SMI). (C) Protein expression levels of three markers of autophagic proteolysis: Beclin, p62, and LC3BI. Protein expression levels were determined by western blot and normalized to Tubulin. Data are presented with IQR box and min‐to‐max whiskers and were compared using Kruskal–Wallis tests with Dunn's post hoc tests. Outliers were excluded using Grubbs' test α = 0.05. Men: black dots, women: white triangles. *P < 0.05, **P < 0.01.
Figure 3
Figure 3
Cancer cachexia affects mitochondrial bioenergetics and structure in pectoralis major muscle. (A) Mitochondrial O2 consumption in permeabilized pectoralis major myofibres from noncachectic patients (NC), patients with mild cachexia (MC), or severe cachexia (SC). O2 consumption related to phosphorylation (state 3—ADP) and to energy waste (state 4—oligomycin); uncoupled state (FCCP); complex 4 (ascorbate and TMPD). (B) Representative observations in TEM (X10000) of longitudinal sections of pectoralis major from NC, MC, and SC patients. (C) Mitochondrial area analysis in TEM. For each patient, four cross‐sections and four longitudinal sections, each on a different myofibre, were used. Median mitochondrial area was calculated on at least 30 mitochondria per section. (D) Quantification of mitochondrial DNA by qPCR. Data are presented with IQR box and min‐to‐max whiskers and were compared using Kruskal–Wallis tests with Dunn's post hoc tests. Outliers were excluded using Grubbs' test α = 0.05. Men: black dots, women: white triangles. *P < 0.05.
Figure 4
Figure 4
Cancer cachexia does not affect mitochondrial biogenesis, fusion, and fission in pectoralis major muscle. (A) mRNA and (B) protein expression of three markers of mitochondrial biogenesis: NRF2, PGC1α, and MTFA, measured in pectoralis major biopsies from noncachectic patients (NC), patients with mild cachexia (MC) or severe cachexia (SC). (C) mRNA and (D) protein expression of three markers of mitochondrial fusion: OPA1, Mfn1, and Mfn2. (E) mRNA and (F) protein expression of three markers of mitochondrial fission: Drp1, Mff, and Fis1. mRNA expression levels were determined by absolute RT‐qPCR, while protein expression levels were determined by Western Blot and normalized to Tubulin. Data are presented with IQR box and min‐to‐max whiskers and were compared using Kruskal–Wallis tests with Dunn's post hoc tests. Outliers were excluded using Grubbs' test α = 0.05. Men: black dots, women: white triangles.
Figure 5
Figure 5
Cancer cachexia does not induce the accumulation of intramyocellular lipid droplets or ER stress in pectoralis major muscle. (A) Lipid droplets area analysis in TEM. For each patient, four cross‐sections and four longitudinal sections, each on a different myofibre, were used. Median area was calculated from all measurable lipid droplets. (B) mRNA expression and (C) protein expression of perilipins measured in pectoralis major biopsies from noncachectic patients (NC), patients with mild cachexia (MC) or severe cachexia (SC). (D) mRNA expression and (E) protein expression of five markers of ER stress: EDEM, ATF4, ATF6, GRP78, and CHOP. mRNA expression levels were determined by absolute RT‐qPCR, while protein expression levels were determined by Western Blot and normalized to Tubulin. Data are presented with IQR box and min‐to‐max whiskers and were compared using Kruskal–Wallis tests with Dunn's post hoc tests. Outliers were excluded using Grubbs' test α = 0.05. Men: black dots, women: white triangles.

References

    1. Fearon K, Strasser F, Anker SD, Bosaeus I, Bruera E, Fainsinger RL, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol 2011;12:489–495.
    1. Bachmann J, Heiligensetzer M, Krakowski‐Roosen H, Büchler MW, Friess H, Martignoni ME. Cachexia worsens prognosis in patients with resectable pancreatic cancer. J Gastrointest Surg 2008;12:1193–1201.
    1. Fearon KC, Voss AC, Hustead DS, Cancer Cachexia Study Group . Definition of cancer cachexia: effect of weight loss, reduced food intake, and systemic inflammation on functional status and prognosis. Am J Clin Nutr 2006;83:1345–1350.
    1. Armstrong VS, Fitzgerald LW, Bathe OF. Cancer‐associated muscle wasting—candidate mechanisms and molecular pathways. Int J Mol Sci 2020;21:9268.
    1. Penna F, Ballarò R, Beltrà M, de Lucia S, García Castillo L, Costelli P. The skeletal muscle as an active player against cancer cachexia. Front Physiol 2019;10:10.
    1. Julienne CM, Dumas J‐F, Goupille C, Pinault M, Berri C, Collin A, et al. Cancer cachexia is associated with a decrease in skeletal muscle mitochondrial oxidative capacities without alteration of ATP production efficiency. J Cachexia Sarcopenia Muscle 2012;3:265–275.
    1. de Castro GS, Simoes E, Lima JDCC, Ortiz‐Silva M, Festuccia WT, Tokeshi F, et al. Human cachexia induces changes in mitochondria, autophagy and apoptosis in the skeletal muscle. Cancer 2019;11:1264.
    1. Marzetti E, Lorenzi M, Landi F, Picca A, Rosa F, Tanganelli F, et al. Altered mitochondrial quality control signaling in muscle of old gastric cancer patients with cachexia. Exp Gerontol 2017;87:92–99.
    1. van der Ende M, Grefte S, Plas R, Meijerink J, Witkamp RF, Keijer J, et al. Mitochondrial dynamics in cancer‐induced cachexia. Biochim Biophys Acta Rev Cancer 2018;1870:137–150.
    1. Anoveros‐Barrera A, Bhullar AS, Stretch C, Esfandiari N, Dunichand‐Hoedl AR, Martins KJB, et al. Clinical and biological characterization of skeletal muscle tissue biopsies of surgical cancer patients. J Cachexia Sarcopenia Muscle 2019;10:1356–1377.
    1. Bhullar AS, Anoveros‐Barrera A, Dunichand‐Hoedl A, Martins K, Bigam D, Khadaroo RG, et al. Lipid is heterogeneously distributed in muscle and associates with low radiodensity in cancer patients. J Cachexia Sarcopenia Muscle 2020;11:735–747.
    1. Stephens NA, Skipworth RJE, Macdonald AJ, Greig CA, Ross JA, Fearon KCH. Intramyocellular lipid droplets increase with progression of cachexia in cancer patients. J Cachexia Sarcopenia Muscle 2011;2:111–117.
    1. Fukawa T, Yan‐Jiang BC, Min‐Wen JC, Jun‐Hao ET, Huang D, Qian CN, et al. Excessive fatty acid oxidation induces muscle atrophy in cancer cachexia. Nat Med 2016;22:666–671.
    1. Barreiro E, Salazar‐Degracia A, Sancho‐Muñoz A, Gea J. Endoplasmic reticulum stress and unfolded protein response profile in quadriceps of sarcopenic patients with respiratory diseases. J Cell Physiol 2019;234:11315–11329.
    1. Bohnert KR, Gallot YS, Sato S, Xiong G, Hindi SM, Kumar A. Inhibition of ER stress and unfolding protein response pathways causes skeletal muscle wasting during cancer cachexia. FASEB J 2016;30:3053–3068.
    1. Roy A, Kumar A. ER stress and unfolded protein response in cancer cachexia. Cancers (Basel) 2019;11:11.
    1. Dolly A, Dumas J‐F, Servais S. Cancer cachexia and skeletal muscle atrophy in clinical studies: what do we really know? J Cachexia Sarcopenia Muscle 2020;11:1413–1428.
    1. Martin L, Birdsell L, MacDonald N, Reiman T, Clandinin MT, McCargar LJ, et al. Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index. J Clin Oncol 2013;31:1539–1547.
    1. Martin L, Senesse P, Gioulbasanis I, Antoun S, Bozzetti F, Deans C, et al. Diagnostic criteria for the classification of cancer‐associated weight loss. JCO 2015;33:90–99.
    1. Mourtzakis M, Prado CMM, Lieffers JR, Reiman T, McCargar LJ, Baracos VE. A practical and precise approach to quantification of body composition in cancer patients using computed tomography images acquired during routine care. Appl Physiol Nutr Metab 2008;33:997–1006.
    1. Shen W , Punyanitya M, Wang Z, Gallagher D, St‐Onge M‐P, Albu J, et al. Total body skeletal muscle and adipose tissue volumes: estimation from a single abdominal cross‐sectional image. J Appl Physiol 2004;97:2333–2338.
    1. Smuder AJ, Roberts BM, Wiggs MP, Kwon OS, Yoo J‐K, Christou DD, et al. Pharmacological targeting of mitochondrial function and reactive oxygen species production prevents colon 26 cancer‐induced cardiorespiratory muscle weakness. Oncotarget 2020;11:3502–3514.
    1. Halle JL, Pena GS, Paez HG, Castro AJ, Rossiter HB, Visavadiya NP, et al. Tissue‐specific dysregulation of mitochondrial respiratory capacity and coupling control in colon‐26 tumor‐induced cachexia. Am J Physiol Regulatory, Integr Comp Physiol 2019;317:R68–R82.
    1. Neyroud D, Nosacka RL, Judge AR, Hepple RT. Colon 26 adenocarcinoma (C26)‐induced cancer cachexia impairs skeletal muscle mitochondrial function and content. J Muscle Res Cell Motil 2019;40:59–65.
    1. Argilés JM, López‐Soriano FJ, Busquets S. Muscle wasting in cancer: the role of mitochondria. Curr Opin Clin Nutr Metab Care 2015;18:221–225.
    1. Dubinin MV, EYu T, Tenkov KS, Starinets VS, Mikheeva IB, Sharapov MG, et al. Duchenne muscular dystrophy is associated with the inhibition of calcium uniport in mitochondria and an increased sensitivity of the organelles to the calcium‐induced permeability transition. Biochim Biophys Acta (BBA) ‐ Molecular Basis Dis 2020;1866:165674.
    1. Strubbe‐Rivera JO, Chen J, West BA, Parent KN, Wei G‐W, Bazil JN. Modeling the effects of calcium overload on mitochondrial ultrastructural remodeling. Appl Sci (Basel) 2021;11:2071.
    1. Sandri M. Protein breakdown in cancer cachexia. Semin Cell Dev Biol 2016;54:11–19.
    1. Zhang Y, Wang J, Wang X, Gao T, Tian H, Zhou D, et al. The autophagic‐lysosomal and ubiquitin proteasome systems are simultaneously activated in the skeletal muscle of gastric cancer patients with cachexia. Am J Clin Nutr 2020;111:570–579.
    1. Stephens NA, Skipworth RJE, Gallagher IJ, Greig CA, Guttridge DC, Ross JA, et al. Evaluating potential biomarkers of cachexia and survival in skeletal muscle of upper gastrointestinal cancer patients. J Cachexia Sarcopenia Muscle 2015;6:53–61.
    1. MacDonald AJ, Johns N, Stephens N, Greig C, Ross JA, Small AC, et al. Habitual myofibrillar protein synthesis is normal in patients with upper GI cancer cachexia. Clin Cancer Res 2015;21:1734–1740.
    1. Bodine SC, Baehr LM. Skeletal muscle atrophy and the E3 ubiquitin ligases MuRF1 and MAFbx/atrogin‐1. Am J Physiol Endocrinol Metab 2014;307:E469–E484.
    1. Martin A, Freyssenet D. Phenotypic features of cancer cachexia‐related loss of skeletal muscle mass and function: lessons from human and animal studies. J Cachexia Sarcopenia Muscle 2021;12:252–273.
    1. Bossola M, Marzetti E, Rosa F, Pacelli F. Skeletal muscle regeneration in cancer cachexia. Clin Exp Pharmacol Physiol 2016;43:522–527.
    1. Okada A, Ono Y, Nagatomi R, Kishimoto KN, Itoi E. Decreased muscle atrophy F‐box (MAFbx) expression in regenerating muscle after muscle‐damaging exercise. Muscle Nerve 2008;38:1246–1253.
    1. Hughes DC, Baehr LM, Driscoll JR, Lynch SA, Waddell DS, Bodine SC. Identification and characterization of Fbxl22, a novel skeletal muscle atrophy‐promoting E3 ubiquitin ligase. Am J Physiol Cell Physiol 2020;319:C700–C719.
    1. Milan G, Romanello V, Pescatore F, Armani A, Paik J‐H, Frasson L, et al. Regulation of autophagy and the ubiquitin‐proteasome system by the FoxO transcriptional network during muscle atrophy. Nat Commun 2015;6:6670.
    1. Marceca GP, Londhe P, Calore F. Management of cancer cachexia: attempting to develop new pharmacological agents for new effective therapeutic options. Front Oncol 2020;10:298.
    1. Johns N, Hatakeyama S, Stephens NA, Degen M, Degen S, Frieauff W, et al. Clinical classification of cancer cachexia: phenotypic correlates in human skeletal muscle. PLoS ONE 2014;9:e83618.
    1. das Neves W, Alves CRR, de Souza Borges AP, de Castro G. Serum creatinine as a potential biomarker of skeletal muscle atrophy in non‐small cell lung cancer patients. Front Physiol 2021;12:625417.

Source: PubMed

3
Prenumerera